(19)
(11) EP 2 419 128 B1

(12) EUROPEAN PATENT SPECIFICATION

(45) Mention of the grant of the patent:
06.06.2018 Bulletin 2018/23

(21) Application number: 10765009.5

(22) Date of filing: 13.04.2010
(51) International Patent Classification (IPC): 
A61K 39/08(2006.01)
(86) International application number:
PCT/US2010/030875
(87) International publication number:
WO 2010/120766 (21.10.2010 Gazette 2010/42)

(54)

ENGINEERED BOTULINUM NEUROTOXIN

MANIPULIERTES BOTULINUMNEUROTOXIN

NEUROTOXINE BOTULINIQUE REMANIÉE


(84) Designated Contracting States:
AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

(30) Priority: 14.04.2009 US 169031 P

(43) Date of publication of application:
22.02.2012 Bulletin 2012/08

(73) Proprietor: Medical College of Wisconsin, Inc.
Milwaukee, WI 53226 (US)

(72) Inventors:
  • BARBIERI, Joseph, T.
    Milwaukee, WI 53226 (US)
  • CHEN, Sheng
    Kow Loon (HK)

(74) Representative: MacLean, Martin Robert 
Mathys & Squire LLP
The Shard 32 London Bridge Street London SE1 9SG
The Shard 32 London Bridge Street London SE1 9SG (GB)


(56) References cited: : 
US-A1- 2004 115 727
US-A1- 2006 211 619
   
  • SHENG CHEN ANDJOSEPH T BARBIERI: "Engineering botulinum neurotoxin to extend therapeutic intervention", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF USA, NATIONAL ACADEMY OF SCIENCE, WASHINGTON, DC; US, vol. 106, no. 23, 1 June 2009 (2009-06-01) , pages 9180-9184, XP008145458, ISSN: 0027-8424, DOI: 10.1073/PNAS.0903111106
  • RAKHI AGARWAL ET AL: "Analysis of Active Site Residues of Botulinum Neurotoxin E by Mutational, Functional, and Structural Studies: Glu335Gln Is an Apoenzyme +", BIOCHEMISTRY, vol. 44, no. 23, 1 June 2005 (2005-06-01), pages 8291-8302, XP055015238, ISSN: 0006-2960, DOI: 10.1021/bi050253a
  • KUKREJA ET AL.: 'Role of two active site Glu residues in the molecular action of botulinum ieurotoxin endopeptidase' BIOCHIMICA ET BIOPHYSICA ACTA vol. 1774, 2007, pages 213 - 222, XP005878417
  • CHEN ET AL.: 'Unique Substrate Recognition by Botulinum Neurotoxins Serotypes A and E.' THE JOURNAL OF BIOLOGICAL CHEMISTRY vol. 281, no. 16, 2006, pages 10906 - 10911, XP008145452
  • MONTAL.: 'Botulinum Neurotoxin: A Marvel of Protein Design.' ANNU. REV. BIOCHEM. vol. 79, 16 March 2010, pages 591 - 617, XP008145457
  • CHEN ET AL.: 'Engineering botulinum neurotoxin to extend therapeutic intervention.' PNAS vol. 106, no. 23, 09 June 2009, pages 9180 - 9184, XP008145458
 
Remarks:
The file contains technical information submitted after the application was filed and not included in this specification
 
Note: Within nine months from the publication of the mention of the grant of the European patent, any person may give notice to the European Patent Office of opposition to the European patent granted. Notice of opposition shall be filed in a written reasoned statement. It shall not be deemed to have been filed until the opposition fee has been paid. (Art. 99(1) European Patent Convention).


Description

CROSS-REFERENCE TO RELATED APPLICATIONS



[0001] This application claims priority to U.S. Provisional Application No. 61/169,031, filed April 14.

STATEMENT REGARDING FEDERALLY-SPONSORED RESEARCH OR DEVELOPMENT



[0002] This invention was made with United States government support awarded by the following agency: NIH grant 1-U54-AI-057153. The United States government has certain rights in this invention.

BACKGROUND OF THE DISCLOSURE



[0003] Clostridium botulinum produces seven different neurotoxins (BoNTs) which differentiated serologically by the lack of anti-serum cross serotype neutralization. BoNTs are the most potent toxins known to humans and are the causative agents of the disease botulism (1). BoNTs exert their action by inhibiting the release of the neurotransmitter acetylcholine at the neuromuscular junction, leading to a state of flaccid paralysis. BoNTs elicit neuronal-specific flaccid paralysis by targeting neurons and cleaving neuron specific SNARE proteins.

[0004] SNARE proteins (Soluble NSF Attachment protein Receptors) are a large superfamily of proteins. The main function of SNARE proteins is to mediate the exocytosis of neurotransmitter molecules to the post-synaptic junction. SNAREs are small, abundant and both vesicle and plasma-membrane bound proteins.

[0005] BoNTs are a 150kDa polypeptide chain comprising a 100kDa heavy chain and a 50kDa light chain linked by a disulfide bond. BoNTs are organized into three functional domains: an N-terminal zinc metalloprotease light chain (LC), a translocation domain (HCT) and a C-terminal receptor binding domain (HCR) (1, 2). The toxic effect of BoNTs (nerve intoxification) is accomplished through the interplay of three key events. One, the carboxy half of the heavy chain is required for receptor-specific binding to cholinergic nerve cells at the nerve-cell membrane. After binding, another portion of the BoNT moves a smaller catalytic domain into the cell, where the catalytic domain binds to and cleaves a neuronal SNARE protein, "intoxicating" the nerve cell, making it impossible to "fire" or send signals. By "catalytic domain" we mean the part of the molecule that triggers the cleavage of the substrate. The toxin is internalized into an endosome through receptor-mediated endocytosis, and the toxin binds the liminal domains of synaptic vesicle-associated proteins upon the fusion of synaptic vesicles with the plasma membrane (3-5). In short, BoNTs are internalized into endosomes and upon acidification, the LC is translocated into the cytoplasm, where SNARE proteins are cleaved (1, 2).

[0006] Mammalian neuronal exocytosis is driven by the formation of protein complexes between the vesicle SNARE, VAMP2, and the plasma member and SNAREs, SNAP 25 and syntaxin 1a (6). There are seven serotypes of BoNTs (termed A-G) that cleave specific residues on one of three SNARE proteins: serotypes B, D, F, and G cleave VAMP-2, serotypes A and E cleave SNAP 25, and serotype C cleaves SNAP 25 and syntaxis 1a (1). Thus, neuronal specificity is based upon BoNT binding to neurons and cleaving neuronal isoforms of the SNARE proteins. For example, BoNT/A cleaves human SNAP25, but not the human non-neuronal isoform SNAP 23 (7, 8). The non-neuronal SNARE isoforms are involved in a divergent cellular processes, including fusion reactions in cell growth, membrane repair, cytokinesis and synaptic transmission.

[0007] The reversible nature of muscle function after BoNTs intoxication that replace toxin-affected nerves with new nerves (10, 11), has turned the BoNTs from a deadly agent to novel therapies for neuromuscular conditions. As early as 1989, BoNT/A was approved by the FDA to treat strabismus, blepharospasm, and hemifacial spasm and then for cervical dystonia, cosmetic use, glabellar facial lines and axillary hyperhidrosis (12). BoNT/A efficacy in dystonia and other disorders related to involuntary skeletal muscle activity, coupled with a satisfactory safety profile, and prompted empirical/off-label use in a variety of secretions and pain and cosmetic disorders (13).

[0008] The clinical use of BoNTs is limited to targeting inflictions affecting neuromuscular activity (12, 13). Elucidation of the structure-function relationship of BoNTs has enabled the design of novel therapies that retarget BoNT to unique neurons and non-neuronal cells. Replacement of BoNT HCR domain with nerve growth factor, lectin from Erythrina cristagalli, or epidermal growth factors enable retargeting of BoNT/A to neuronal or non-neuronal cells such as nociceptive afferents and airway epithelium cells (14-16). However, the selective cleavage of neuronal specific SNARE proteins by BoNT has limited development of novel therapies in these non-neuronal systems. Prerequisite to develop novel therapies requires the retargeting of the catalytic activity of the BoNTs to non-neuronal SNARE isoforms.

[0009] Accordingly, a need exists for an engineered BoNT that cleaves non-neuronal SNARE proteins and methods of use thereof.

SUMMARY



[0010] The invention is defined by the claims. Those aspects/instances of the present disclosure which constitute the invention are defined by the claims.

[0011] In one instance, the present disclosure provides a modified BoNT/E catalytic domain, wherein light chain residue 224, or a residue corresponding to residue 224, has been altered. In one instance, the residue 224 has been altered to be aspartic acid or glutamic acid. The modified catalytic domain cleaves SNAP23 but does not cleave SNAP29 or SNAP47.

[0012] In an alternate instance, the modified catalytic domain additionally comprise a target molecule useful in a protein delivery system.

[0013] In an alternate instance, the present disclosure provides an engineered botulinum neurotoxin E or botulinum neurotoxin light chain or botulinum toxin catalytic domain comprising a modified BoNT/E catalytic domain, wherein light chain residue 224 has been altered.

[0014] In an alternate instance, the present disclosure provides a method of treating a subject in need of botulinum toxin therapy, comprising the step of administering a therapeutically effective amount of a modified BoNT/E catalytic domain, wherein light chain residue 224 has been altered, to the subject. The subject in need of botulinum toxin therapy may suffer from, without limitation, asthma, CF, chronic obstructive pulmonary, gastric acid efflux and inflammation, immune disorders with a cytokine component or cancers with a cytokine component.

[0015] While multiple instances are disclosed, still other instances of the present disclosure will become apparent to those skilled in the art from the following detailed description.

[0016] The detailed descriptions are to be regarded as illustrative in nature and not restrictive.

BRIEF DESCRIPTION OF THE FIGURES



[0017] 

Figure 1. K185 of human SNAP23 contributes to the substrate recognition by BoNT/E. (a) Substrate recognition by LC/E. Two subsites in SNAP25 contribute to substrate binding "B" (Km) and catalysis "AS" (kcat), where the P3, P2, and PI' residues contribute to recognition by LC/E. (b) Sequence alignment of human SNAP25 (SN25) and human SNAP23 (SN23). (c) (upper panel) modeled complex structure of LC/ESNAP25 predict the recognition of P site residues of SNAP25 by LC/E. (lower panel) modeled complex structure of LC/E(K224D)-SNAP23 predict the recognition of P site residues of SNAP23 by LC/E(K224D). Models were generated by SWISS-MODEL, using LC/E crystal structure (PDB:3d3x), and images were generated in PyMol.

Figure 2. Cleavage of SNAP23 by LC/E(K224D). (a) Five µm SNAP23 was incubated with indicated amounts of LC/E(K224D) and subjected to SDS-PAGE (stained gel is shown in insert, SNAP23(152-211) is designated (SN23(152-211)) and the cleavage product SNAP23(152-186) is designated*. %SNAP23 cleavage was determined by densitometry. (b) Kinetic constant for LC/E to cleave SNAP25 and LC/E(K224D) to cleave SNAP23.

Figure 3. Site of SNAP23 cleavage by LC/E(K224D) (a) Five µm SNAP23 was incubated with 2 µm of LC/E(K224D) and subjected to MALDI-TOF Mass Spectrometry. Intensity (100%) on the y-axis was set to the 2812.5 band and the x-axis represents mass-to-charge units, m/z. (b) SNAP23(I187D) was incubated with the indicated amounts of LC/E(K224D) and subjected to SDS-PAGE. The Coomassie stained gel is shown with the migrations of LC/E(K224D and SNAP23(I187D) indicated on the left.

Figure 4. Sequence alignment and substrate specificity of LC/E(K224D) and Wt- LC/E on SNAP25 isoforms. (a) Alignment of SNAP23a,b, SNAP25a,b, SNAP29 and SNAP47 (ClustaIW2) in the regions corresponding to SNARE proteins that interact with the binding region and active sites region of LC/E. Indicated are conserved residues (*) and similar residues (:, .) among the SNAP25 isoforms. Cleavage site of SNAP25 by LC/E (arrow) and P site resides are indicated. Linear velocity assays of LC/E(K224D) (b) and Wt-LC/E (c) with the indicated isoforms of SNAP25. Five µm SNAP25 isoform was incubated with the indicated amounts of LC, subjected to SDS-PAGE and gels were stained with Coomassie. The amount of SNAP25 isoform cleavage was determined by densitometry.

Figure 5. LC/E(K224D) cleaves SNAP23 and inhibits mucin and IL-8 secretion in TGF-α stimulated HeLa cells. (A) GFP-LC/E(K224D) or GFP-Wt-LC/E were transfected into HeLa cells. After 24h, cell lysates were prepared and separated by SDS-PAGE and cleavage of SNAP23 was measured by western-blotting using anti SNAP-23 mouse monoclonal antibody. (B, C). HeLa cells were transfected with DNA encoding GFP-LC/E(K224D) or GFP-Wt-LC/E. After 24h, cells were washed with serum free MEM medium twice and secretion was induced by the addition of serum free MEM medium supplemented with 20ng/ml of TNF-α. After 36h, supernatant were collected and assayed for mucin and IL-8 secretion, using an ELISA format. The amount of mucin and IL-8 secreted in controls cells was adjusted to 1.0 and used as a reference for cells treated with TNF-α.

Figure 6. Recombinant LC/E(K224D) cleaves SNAP23 and inhibits mucin and IL-8 secretion in TGF-α stimulated HeLa cells. HeLa cells were treated with digitonin and then incubated with His-LC/E(K224D) or His-Wt-LC/E (3-Xflag tagged proteins). After an overnight incubation, cells were washed and then incubated with serum free MEM media supplemented with 20 ng/ml of TNF-α for 36 h when cell supernatants were collected and cell lysates were prepared. (a) Cell lysates were subjected to SDS-PAGE and LC/E expression and SNAP23 cleavage was measured by Western blot, using α-3Xflag antibody and α-SNAP23 antibody, respectively; * indicates migration of the SNAP23 cleavage product. Culture supernatants were assayed IL-8 (b) and mucin (c) secretion, using an ELISA, using 1.0 as a reference for cells treated with recombinant LC/E.


DETAILED DESCRIPTION


I. IN GENERAL



[0018] In the specification and in the claims, the terms "including" and "comprising" are openended terms and should be interpreted to mean "including, but not limited to.... " These terms encompass the more restrictive terms "consisting essentially of" and "consisting of."

[0019] As used herein and in the appended claims, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. As well, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. It is also to be noted that the terms "comprising", "including", "characterized by" and "having" can be used interchangeably.

[0020] Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this disclosure belongs.

[0021] All references cited in this specification are to be taken as indicative of the level of skill in the art. Nothing herein is to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

II. THE DISCLOSURE



[0022] The present disclosure provides novel engineered botulinum neurotoxins (BoNTs) that cleave non-neuronal SNARE proteins and methods of use thereof.

[0023] BoNTs are effective therapeutics for a variety of neurological disorders, such as strabismus, blepharospasm, hemificial spasm, and cervical dystonia, due to the toxin's tropism for neurons and specific cleavage of neuronal SNARE proteins. Modifying BoNTs to bind non-neuronal cells requires retargeting the catalytic activity of BoNTs to non-neuronal SNARE isoforms to provide effective non-neuronal therapies.

[0024] Here, we extend the substrate specificity of BoNT/E by engineering a catalytic derivative that cleaves the non-neuronal SNARE protein, SNAP23, as a platform for novel methods of treating non-neuronal human secretory diseases. By "non-neuronal human secretory diseases, we mean, for example, diseases or conditions where excessive airway mucus secretion, mucus hypersecretion, may cause mucus accumulation that is associated with human clinical conditions such as asthma and chronic obstructive pulmonary disease where mucus accumulation contributes to respiratory disease. Specifically, we now report for the first time the engineering of a BoNT/E derivative that cleaves SNAP23, a non-neuronal SNARE protein.

[0025] SNAP23 mediates vesicle-plasma membrane fusion processes, including secretion of airway mucus, antibody, insulin, gastric acids, and ions. The mutated BoNT/E light chain LC/E(K224D) of the presentdisclosure showed extended substrate specificity to cleave SNAP23, and the natural substrate, SNAP25, but showed no specificity for, and did not cleave, SNAP29 or SNAP47. Upon direct protein delivery into cultured human epithelial cells, LC/E(K224D) cleaved endogenous SNAP23, which inhibited secretion of mucin and IL-8. These studies show for the first time the feasibility of genetically modifying BoNTs to target a non-neuronal SNARE protein for novel methods of treating human hyper-secretion diseases, such as asthma and chronic obstructive pulmonary disease.

[0026] In one instance, the present disclosure is a preparation of an engineered catalytic domain of botulinum neurotoxin E that is capable of specifically cleaving SNAP23. Preferably, the toxin cleaves SNAP25 but not SNAP29 or SNAP 47. The LC/E(K224D) can cleave both SNAP25 and SNAP23, but at ∼10 fold lower than Wt-LC/E can cleave SNAP25.

[0027] In a preferred instance, the modified botulinum neurotoxin E of the present disclosure has a mutation at light chain residue 224 or a residue corresponding to residue 224. By "residue 224" we mean the lysine at position numbered 224 of SEQ ID NO: 7. SEQ ID NO: 7 is the protein conversion of the DNA sequence of SEQ ID NO: 1 absent the first methionine residue. By "residue corresponding to residue 224" we mean the lysine within the motif of residues 210-240 of SEQ ID NO: 7. Specifically, we mean the highlighted lysine within residues MHELIHSLHGLYGAKGITTKYTITQKQNPLI (SEQ ID NO: 8). Other botulinum type E subtypes have this same corresponding residue and motif, although the numbering may not be identical among subtypes. However, the motif residues will be at least 90% corresponding to the motif of SEQ ID NO: 8.

[0028] One may find an exemplary sequence of botulinum toxin E subtype Beluga light chain at GeneBank with accession number X62089 (SEQ ID NO: 1). We anticipate that other subtypes of serotype E could be used as template for engineering LC that can cleave SNAP23.One would modify residue numbered 224 of any botulinum toxin E subtype in the same manner as disclosed within the present disclosure.

[0029] In a preferred instance, the present disclosure is a preparation of a modified botulinum toxin E with a mutation at a light chain residue 224 which cleaves SNAP23. Another instance of the present disclosure is a preparation of botulinum neurotoxin E light chain with a mutation at light chain residue 224 which cleaves SNAP23. Another instance of the present disclosure is a preparation of botulinum neurotoxin catalytic domain (residues 1 through 400) with a mutation at light chain residue 224 which cleaves SNAP23. Another instance of the present disclosure is a truncated fragment of the catalytic domain, comprising at least residues 1 to 390, which comprises a modified residue 224 which cleaves SNAP23. By "modified catalytic domain" we mean to include all forms, including humanized forms, of the catalytic domain of L/C BoNT with a modification at residue 224.

[0030] Another preferred instance is a mutated botulinum neurotoxin E light chain or modified catalytic domain fused to another peptide, as described below, for appropriate therapeutic methods.

[0031] In another instance, the present disclosure is a DNA sequence encoding the engineered botulinum neurotoxin or the modified catalytic domain described herein.

[0032] An exemplary reference to the sequences of SNAP23, SNAP25, SNAP29 and SNAP47 can be found at GeneBank with accession numbers CR457212 (SEQ ID NO: 2), NM_130811 (SEQ ID NO: 3), CR456582 (SEQ ID NO: 4) and BC011145 (SEQ ID. NO: 5).

[0033] In other instances of the disclosure, other mutations of residue 224 would also be suitable. The mutation within LC/E that is being protected is K224D, which recognizes the P2 residue of SNAP23, which is a lysine. We anticipate that a glutamic acid mutation at K224 would also yield a functional LC/E that can cleave SNAP23. We generated a K224A mutation that has the ability to cleave both SNAP23 and SNAP25, but with less efficiency than K224D, which indicates that charge and size of the R-group influence cleavage efficiency. Thus, we anticipate that other residue replacements, such as replacing the lysine at residue 224 of SEQ ID NO: 7 with glutamic acid, may also provide a mutated LC/E with the ability to cleave SNAP23.

[0034] In other instances, we further anticipate that additional residue replacements, alone or in combination with the mutation of K224D of the present invention, may yield a LC/E that has the ability to cleave SNAP 23 and not SNAP 25.

[0035] Methods of Treatment. In other instances, the present disclosure provides novel methods of treating a subject requiring treatment with a botulinum toxin. In one instance, the present disclosure provide methods of treating a subject suffering from a human secretory disease by administering a therapeutically effective amount of the modified catalytic domain as described above.

[0036] By "subject" we mean any person requiring treatment with botulinum toxin. By "treating" or "treatment", we mean the management and care of a subject for the purpose of combating the disease, condition, or disorder. The terms embrace both preventative, i.e., prophylactic, and palliative treatment. Treating includes the administration of a compound of present disclosure to prevent the onset of the symptoms or complications, alleviating the symptoms or complications, or eliminating the disease, condition, or disorder. In one instance, we envision the method of the presentdisclosure reducing symptoms by at least 20 to 50 percent. We envision treatment occurring on a regular basis until symptoms are reversed. For instance, in one instance, treatment would occur daily, weekly or monthly, as needed.

[0037] By "therapeutically effective amount" we mean amount of a compound that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. The "therapeutically effective amount" will vary depending on the compound, the disease state being treated, the severity or the disease treated, the age and relative health of the subject, the route and form of administration, the judgment of the attending medical or veterinary practitioner, and other factors as known to one of skill in the art (40). This method would involve administering to the subject the modified catalytic domain of the engineered botulinum toxin E of the present disclosure. By "administering" we mean delivering the engineered BoNT of the present disclosure to the subject. Although the preferable form of the catalytic domain is the botulinum toxin E light chain, the catalytic domain may include forms that are shorter than the light chain, such as the C-terminal truncation mutants of the catalytic domain to residues ∼1-390 residues (41), and forms that are larger than the light chain and include the translocation domain or fragments of the translocation domain up to ∼residue F1200 which includes the N terminus of the HCR domain. In one instance, we have engineered a 1-400 which has good solubility and activity.

[0038] In one instance, the catalytic domain is complexed, either through covalent bonds or attached in some other way, such as cross-linking, to a targeting molecule (targeting system). The targeting molecule is adapted to target the toxin to a cell-surface receptor of interest.

[0039] As described above, we envision several forms of the catalytic domain as being effective potential delivery platforms for the K224D mutation. For instance, protein solubility may vary with the application and the chimera that is engineered, therefore different sized catalytic domains may be more useful in specific applications. We anticipate that the amount of effective delivery platform will be similar to the amounts of BoNT used in clinical therapy and that the amount of effective delivery can be fractional based upon the catalytic nature of LC/E(K224D).

[0040] In other instances, a suitable delivery system will, in one instance, target specific cell types or cell surface receptors that are internalized and deliver LC/E into the cytoplasm, such as an antibody or a growth factor. Potential cell surface receptors that can be targeted include tissue specific growth factor receptors, which could vary with the disease that is targeted. For example, one of skill would understand based on materials known to the field how to target CD22 in hairy cell leukemia (42).

[0041] Alternatively, a lipid-based directed protein delivery system could be used to deliver LC/E directly into the host cytosol. For instance, a targeted-liposome delivery which delivers either the catalytic domain protein directly or DNA encoding the LC/E(K224D) protein (39) are also effective methods of delivering the engineered BoNT/E of the present disclosure to treat various diseases. For instance, lipid based delivery system that assemble nano-particles for efficient delivery and reduced immunogenicity should prove useful as targeting vehicles (43). While the different delivery methods may not have different advantages based on the disease being targeted, the specificity of the delivery system will be a critical factor in selecting a specific delivery system.

[0042] To optimize the potency, we envision humanizing the modified catalytic domain using one of several possible approaches known to the art (34, 35).

[0043] Several delivery systems are envisioned for the therapeutic delivery of LC/E(K224D), or another suitable toxin, to a subject requiring treatment for, for instance, a non-secretory or hyper-secretory disease. Example delivery systems include, without limitation, single chain protein chimeras where the modified catalytic domain, preferably LC/E(K224D), is fused either at the DNA level or by protein-receptor cross-linking (36) or bipartate protein delivery systems where the catalytic domain, preferably LC/E(K224D), is linked to a fusion composed of a di-protein delivery system (37, 38). For example, the gene encoding LC/E(K224D) could be genetically fused to the gene encoding the epidermal growth factor to target non-small-cell lung cancer.

[0044] The table below describes some of the appropriate uses for the engineered toxin of the presentdisclosure and appropriate targets for the targeting domain.
TABLE 1: Receptor/Disease
LC/E(K224D)-Receptor Disease
Lung epithelium cell specific receptor Asthma, CF, chronic obstructive pulmonary disease
Gastric specific receptor Gastric acid efflux and inflammation
Mast cell specific receptor Allergic rhinitis, Hemophagocytic lymphohistiocytosis, and Chronic urticaria
Cancer specific receptor Renal cell carcinoma, Nonsmall cell lung cancer, and gastric cancer, Epithelial ovarian cancer, and Estrogen receptor (ER)-positive breast cancer


[0045] Kits. In an alternate instance of the disclosure, a kit for treating a subject with the modified catalytic domain of the present disclosure is provided. In one instance, the kit comprises a form of the engineered BoNT of the present disclosure and instructions for use. In one instance, the modified catalytic domain of the present disclosure is formulated, delivered and stored for use in physiologic conditions. In a preferred instance, the kit also comprises a targeting system. The modified catalytic domain is either already attached to the targeting system or the kit contains the targeting system with instructions for attachment. In alternate instances, the kit comprises DNA encoding LC/E(K224D) that can be used to engineer fusion proteins to specific tissue specific targeting molecules.

[0046] By "instructions for use" we mean a publication, a recording, a diagram, or any other medium of expression which is used to communicate the usefulness of the disclosure for one of the purposes set forth herein. The instructional material of the kit can, for example, be affixed to a container which contains the present disclosure or be shipped together with a container which contains the disclosure. Alternatively, the instructional material can be shipped separately from the container or provided on an electronically accessible form on an internet website with the intention that the instructional material and the biocompatible hydrogel be used cooperatively by the recipient.

III. EXAMPLES



[0047] The following examples are offered for illustrative purposes only and are not intended to limit the scope of the present invention in any way. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and the following examples and fall within the scope of the appended claims.

A. METHODS AND MATERIALS.



[0048] Molecular modeling. Molecular modeling was performed using SWISS-MODEL. The structure of the LC/E-SNAP25 (146-202) complex was obtained as described (19), using the crystal structure of LC/E (PDB:3d3x). The structure of LC/E(K224D)-SNAP23 was modeled using LC/E-SNAP25 complex structure as a template, using PyMol™. Data presented are the average of experiments performed at least 3 times.

[0049] Plasmid construction and protein expression. BoNT LC/E expression vector was constructed by amplifying DNA encoding LC/E (1-400) of Clostridium botulinum serotype E Beluga (SEQ ID NO: 1) and subcloning into pET-15b. For transfection experiments, LC/E(1-400) was also subcloned into pEGFP vector to generate an EGFP-LC/E (1-400) fusion protein expressed under the CMV promoter. Expression vectors for SNAP23(152-211), SNAP29(202-259) and SNAP47(406-464), the protein equivalents of SNAP25(145-206), were constructed by PCR amplifying and cDNA template: human SNAP23 (ATCC 2900640, SEQ ID NO:2), SNAP29(ATCC 10700609, SEQ ID NO: 4) and SNAP47(ATCC 10468826, SEQ ID NO: 5) and subcloning into pGEX-2T. Site-directed mutagenesis was performed using QuickChange (Stratagene). Protein expression and purification were performed as previously described (32).

[0050] Cleavage of SNARE protein by LC/E and LC/E(K224D). Cleavage of SNARE protein was performed as described.

[0051] Linear velocity reaction: Reactions contained (10 µl): 5 µM human SNARE proteins, 10mM Tris-HCl (ph 7.6) with 20 mM NaCl, and the indicated amounts of LC/E and LC/E(K224D). Reactions were incubated for 10 min at 37°C, subjected to SDS-PAGE and gels were stained with Coomassie. The amount of SNARE protein cleavage was determined by densitometry.

[0052] Kinetic parameters: Km and kcat determinations were made for Wt-LC/E and LC/E derivatives using SNAP25 isoforms. LC concentrations were adjusted to cleave <10% substrate at several concentrations of substrate (1.5∼18 µM SNARE protein). Reactions were carried out at 37°C for 10 min, subjected to SDS-PAGE and the amount of cleaved product was calculated by densitometry. Reaction velocity versus substrate concentration was fit to the Michaelis-Menten equation, using Lineweaver-Burk plots, using SigmaPlot IX (Chicago, IL).

[0053] LC/E and LC/E(K224D) activity in human cultured epithelial cells. HeLa cells were cultured in 6 well plates in MEM supplemented with 10% newborn calf serum. Sub-confluent cells were transfected with 0.5 or 1.0 µg of indicated plasmid using Lipofectamine LTX (GIBCO/BRL).

[0054] Protein delivery. Protein delivery was performed as described (33) with modification. HeLa cells were permeabilized with 1 ml/well of permeabilization buffer containing 30 µM digitonin for 7 min and the incubated in permeabilization buffer with and without the indicated LC.

[0055] Protein Secretion Assays: After an overnight incubation, transfected and protein delivered cells were incubated in 2m1 serum free MEM containing 20 ng/ml of TNF-α. After 36h, 1.5 ml of supernatant was collected, centrifuged at 13,000 g for 1 min, and assayed for secreted mucin and IL-8, using ELISA. Supernatants (150 µl) were mixed with 50 µl of 0.2 M Na2CO3 (pH 9.6) and added to 96-well plates and incubated overnight at 4°C. Plates were washed and locked with 50 mM Na2CO3 (pH 9.6) buffer containing 1% (w/v) BSA. Plates were washed and incubated with 100 µl α-mucin IgG (1/200 dilution, Abcam) or α-IL-8 IgG (1/200 dilution, Abcam) for 1h at RT. Plates were washed 3 and incubated with α-mouse Horse Radish Peroxidase-conjugate antibody (1:10,000 dilution, Pierce) for 1h at RT. Plates were washed and developed with 100 µl of Ultra-TMB (Pierce) for 20 min at RT and quenched with 100 µl of 1M H2SO4. A450 was expressed as fraction relative to secreted mucin or IL-8 in control supernatants.

[0056] Cleavage of SNAP23: lysates from cells incubated with TNF-α for 36h were assayed for LC/E and LC/E(K224D)-mediated a cleavage of endogenous SNAP23, using α-SNAP23 mouse IgG (Abcam, Cambridge, MA) by Western blot analysis (19).

[0057] Delivery Systems. The mutated BoNT was administered to a subject using the following delivery methods:

Target-specific Cell Receptor: One skilled in the art of molecular biology would be able to engineer chimeras of LC/E(K224D) fused to targeting molecules, using polymerase chain reaction-like protocols.

Lipid-based: One skilled in the use of lipid based delivery systems would be able to develop lipid LC/E(K224D) ratios for the efficient internalization of the LC by utilizing a lipid to protein matrix.



[0058] Methods of Treatment. The engineered BoNT of the present disclosure was administered to a subject to treat, in one instance, hyper-secretory and non-secretory diseases. One of skill would identify a subject for treatment with the engineered BoNT, administer the treatment, monitor the results, and determine the effectiveness of the treatment, following strategies utilized for the development of immunotoxin therapy in the treatment of hairy cell leukemia.

B. RESULTS.



[0059] Previous studies identified residues 167-186 as the minimal, optimal peptide of SNAP25, a 206 amino acid protein for LC/E in vitro cleavage (19). SNAP25 (167-186) comprises two sub-sites that include a substrate binding "B" region and an active site "AS" region (Fig. 1a). LC/E recognizes the P3 residue to facilitate alignment of the P2 and P1' residues of SNAP25. The S1' pocket of LC/E is formed by F191, T159, and T208 with hydrophobic interactions between F191 of LC/E and the P1' residue I181 of SNAP25 (20). The basic S2 pocket contains K224, which recognizes the P2 residue D179, through a predicted salt bridge. Docking the P2 and P1' residues of SNAP25 into the active site pockets of LC/E aligns the scissile bond for cleavage (19, 20).

[0060] BoNT/E was known as not cleaving human SNAP23 (8), providing a framework of defining SNAP isoforms specificity of the BoNTs. Many of the residues that contributed to LC/E recognition of SNAP25 were conserved in human SNAP23, except T173/A179, D179/K185, M182/T188 and E183/D189, respectively (Fig. 1b). T173 in SNAP25 played only a limited contribution for LC/E substrate recognition (20) and only main chain interactions of M182-D186 contributed to LC/E substrate recognition. Thus, the T173/A179, D179/K185, M182/T188 and E183/D189 differences between SNAP25 and SNAP23 did not appear to contribute to inability of cle to cleave SNAP23. In contrast, the P2 residue of SNAP25, D179, is recognized by the basic S2 pocket of cle via the basic residue, K224, which contributes to cle substrate recognition (Fig. 1c, upper panel). Accordingly, the inventors examined whether the salt bridge between K224 of LC/E and D179 of SNAP25 contributes the ability of LC/E to cleave SNAP25 and that charge repulsion between K224 of LC/E and the P2 residue of SNAP23, K185, contributes to the inability ofLC/E to cleave SNAP23. To test this hypothesis, a point mutation, K224D, was introduced into LC/E and tested for the ability to cleave human SNAP23 (Fig. 1c, lower panel).

[0061] LC/E(K224D) cleaved human SNAP23 with a Km of ∼ 3 µM and kcat of ∼17 S-1 (Fig. 2), with 2-fold of the Km and 5-fold of the kcat of LC/E for the cleavage of human SNAP25. The specific activity for the cleavage of SNAP23 by LC/E(K224D) was similar to the cleavage of VAMP-2 by the B serotype of BoNT and ∼10 fold faster for the cleavage of VAMP-2 by tetanus toxin (21, 22). The site that LC/E(K224D) cleaved SNAP23 was identified by MALDI-TOF MS were a major peak with an m/z value of 2812.5 was identified in a reaction mixture that contained SNAP23 and LC/E(K224D) (Fig. 3a), corresponding to the C-terminal 25 amino acid of human SNAP23, IKRITDKADTNRDRIDIANARAKKLIDS (SEQ ID NO: 6). This indicated that LC/E(K224D) cleaved SNAP23 between 186R-I187. The determination that LC/E(K224D) did not cleave SNAP23(I187D) (Fig. 3b) supported that LC/E(K224D) cleaved human SNAP23 between residues 186R-I187.

[0062] SNAP25 isoforms include SNAP25a, SNAP25b, SNAP23a, SNAP23b, SNAP29 and SNAP47 (23, 24). SNAP23 and SNAP25 mediate synaptic membrane fashion in non-neuronal and neuronal cells, respectively, while SNAP29 and SNAP47 have not been implicated in membrane fusion events. SNAP29 was shown to inhibit SNARE disassembly and was implicated in synaptic transmission (25). While the function of SNAP47 is not clear, SNAP47 can substitute for SNAP25 in SNARE complex formation and proteoliposome fashion. The substrate specificity of LC/E(K224D) on SNAP25 isoforms including SNAP23a, SNAP25b, SNAP29 and SNAP47 were tested. SNAP23b and SNAP25a were not tested because the a-b isoforms of SNAP23 and SNAP25 were identical and the LC/E substrate recognition region (Fig. 4a). LC/E(K224D) showed similar activity on both SNAP23 and SNAP25 (Fig. 4b), but did not cleave SNAP29 and SNAP47. Wt-LC/E cleaved SNAP25 (Fig. 4c), but not the other SNAP25 isoforms. The specificity of another LC/E K224 mutation (K224A) on SNAP23 and SNAP25 was also characterized. LC/E(K224A) cleaved SNAP23 and SNAP25 with similar efficiencies, but at a slower rate than LC/E(K224D) (data not shown).

[0063] Next, the ability of LC/E(K224D) to cleave endogenous SNAP23 in HeLa cells was tested. While a role for SNAP23 in constitutive exocytosis is not apparent (26), SNAP23 contributes to regulated exocytosis (27). Transfection of ∼60% of HeLa cell population with LC/E(K224D) resulted in the cleavage ∼45% of the SNAP23, while SNAP23 cleavage was not detected when HeLa cells were transfected with Wt-LC/E or a no plasmid control (Fig. 5). This indicated that LC/E(K224D), but not Wt-LC/E, cleaved endogenous SNAP23 in cultured cells. The effect of SNAP23 cleavage on HeLa cell secretion was tested in LC/E(K224D)-transfected HeLa cells by analyzing TNF-α- mediated mucin and IL-8 secretion. Control HeLa cells secreted mucin and IL-8 upon addition of TNF-α, while LC/E(K224D)-transfected HeLa cells showed reduced mucin and IL-8 secretion (Fig. 5b and c). The inhibition was specific, since Wt-LC/E-transfected HeLa cells showed the same amount of mucin and IL-8 secretion as control cells and did not cleave endogenous SNAP23 (Fig. 5).

[0064] To test the feasibility of utilizing LC/E(K224D) as a protein therapy, recombinant LC/E(K224D) was delivered into HeLa cells, using digitonin. Recombinant LC/E(K224D) cleaved endogenous SNAP23 (Fig. 6), which inhibited TNF-α-mediated mucin and IL-8 secretion (Fig. 6b and c). Digitonin treatment also delivered Wt-LC/E into HeLa cells, but Wt-LC/E-treated HeLa cells, did not show detectable inhibition of mucin and IL-8 secretion and did not cleave endogenous SNAP23 (Fig. 6). This supports a role for SNAP23 in regulated exocytosis pathways in epithelial cells and indicates the utility of LC/E(K224D) as a research tool to study SNAP23-regulated exocytosis (27).

C. DISCUSSION.



[0065] Understanding of substrate specificity of botulinum neurotoxins has enabled the engineering of a novel light chain derivative of BoNT/E with extended substrate specificity, providing a proof of principle to extend the clinical potential of BoNT therapy beyond neurological applications. While airway mucus protects the epithelial lining by entrapping and clearing foreign debris, bacteria, and viruses from the airway by ciliary movement, a process termed mucociliary clearance (17, 18), excessive airway mucus secretion, mucus hypersecretion, may cause mucus accumulation that is associated with human clinical conditions such as asthma and chronic obstructive pulmonary disease where mucus accumulation contributes to respiratory diseases. Mucus secretion is a regulated process coordinated by several molecules, including SNARE proteins, myristoylated alanine-rich C kinase substrate (MARCKS), and Munc proteins, which coordinate the docking of mucin containing vesicles with the secretory cell plasma membrane for exocytosis (17, 18). Targeting SNAP23 by a substrate modified BoNT may reduce the secretion processes of hypersecretion syndromes. A SNAP23-specific BoNT may also be targeted for other therapeutic applications that include diabetes and inflammatory and immune disorders which also include a hypersecretory component (28, 29).

[0066] Alignment and biochemical analyses allow prediction of the mechanism for the catalytic activity of Wt-LC/E and LC/E(K224D) on SNAP25 isoforms. The low overall homology within the active site regions of SNAP29 and SNAP47 to SNAP25 and the lack of an isoleucine at the P1' site explain the inability of Wt-LC/E and LC/E(K224D) to cleave SNAP29 and SNAP47. In contrast, the overall homology between SNAP23 and SNAP25 is high, except at the P2, P2' and P3' residues with the most dramatic change at the P2 residue where SNAP25 contains an aspartate and SNAP23 contains a lysine. Thus, one reason for the inability of Wt- LC/E to cleave SNAP23 may be due to the electrostatic repulsion of the P2 residue lysine within SNAP23 by K224 of LC/E. This may destabilize the S2 pocket and affect alignment of the P1' residue into the S1' pocket. The ability of LC/E(K224D) to cleave SNAP23 may be due to the introduction of a salt bridge between the P2 residue Lys of SNAP23 and the mutated S2 pocket residue D224. LC/E(K224D) also retained the ability to cleave SNAP25, although at a rate that was ∼10 fold slower than Wt-LC/E. This suggests that the repulsion between the P2 residue aspartate of SNAP25 and the mutated S2 pocket residue D224 was not sufficient to inhibit sessile bond cleavage by LC/E(K224D).

[0067] Since LC/E(K224A) cleaved SNAP25 and SNAP23, but at a reduced rate relative to LC/K224D, both charge and size of the R-group at residue 224 contribute to optimal scissile bond cleavage. Overall, the biochemical properties of LC/E and LC/E-K224 derivatives are consistent with P2 residue-S2 pocket residue interactions contributing to the efficiency of sessile bond cleavage, while not binding SNAP29 and SNAP47. In contrast, the overall homology between SNAP23 and SNAP25 is high, except at the P2, P2' and P3' residues with the most dramatic change at the P2 residue where SNAP25 contains an aspartate and SNAP23 contains a lysine. Thus, one reason for the inability of Wt-LC/E to cleave SNAP23 may be due to electrostatic repulsion of the P2 residue lysine within SNAP23 by K224 of LC/E. This may destabilize the S2 pocket and affect alignment of the PI' residue into the S1' pocket.

[0068] The ability of LC/E(K224D) to cleave SNAP23 may be due to the introduction a salt bridge between the P2 residue Lys of SNAP23 and the mutated S2 pocket residue D224. LC/E(K224D) also retained the ability to cleave SNAP25, although at a rate that was ∼10 fold slower than Wt-LC/E. This suggests that the repulsion between the P2 residue aspartate of SNAP25 and the mutated S2 pocket residue D224 was not sufficient to inhibit sessile bond cleavage by LC/E(K224D). Since LC/E(K224A) cleaved SNAP25 and SNAP23, but at a reduced rate relative to LC/EK224D, both charge and size of the R-group at residue 224 contribute to optimal scissile bond cleavage. Overall, the biochemical properties of LC/E and LC/E-K224 derivatives are consistent with P2 residue-S2 pocket residue interactions contributing to the efficiency of sessile bond cleavage. While the ability of native LC/E to bind SNAP23 has not been determined, kinetic values for LC/E and SNAP25 and LC/E(K224D) and SNAP23 are within 2-fold, indicating similar binding affinities.

[0069] Alignment of SNAP25 and SNAP23 within the LC/E binding region (Fig.1) is nearly identical with 7 of 8 residues identical and the non-identical pair being T:A; a conserved substitution pair, which also supports similar binding affinities of LC/E for SNAP25 and SNAP23. SNARE proteins are key proteins in membrane fusion and trafficking within neuronal secretary pathways (9). The use of BoNT has contributed to the understanding vesicle fusion and neurotransmitter release mechanisms in neuronal cells. The ability of a BoNT derivative to cleave non-neurological SNAREs may provide a useful tool to investigate intracellular vesicular trafficking and the mechanism of membrane fusion in nonneuronal systems.

[0070] Although BoNT/A could be considered the logical serotype to be engineered for novel applications due to its wide clinical applications, analysis of the mechanisms of SNAP25 recognition indicate that LC/A requires a longer substrate for optimal SNAP25 recognition with a greater number of residue interactions than LC/E (20). The less-complex SNAP25-LC/E interactions make BoNT/E amenable for engineering to modify substrate recognition. In addition, alignment of human SNAP25 and SNAP23 showed that these proteins had a high level of homology at the P3 and PI' sites that are involved in SNAP25 recognition by LC/E. Thus, BoNT/E is a useful platform to engineer mutations that effect SNARE protein recognition. The successful delivery of LC/E(K224D) into cells to inhibit IL-8 and mucin secretion supports a role for LC/E(K224D) as a research tool and also shows the potential for therapy to regulate human hypersecretion diseases such as asthma and inflammatory diseases. The therapeutic specificity of LC/E(K224D) would be based upon the receptor binding component, as described for toxin chimeras, such as diphtheria toxin A fragment-IL2 (30) and Exotoxin A fragment-IgG variable region fragment (31). In conclusion, the current study shows proof of principle for altered substrate specificity to extend the application of BoNTs beyond neurological inflictions.

[0071] It should be noted that the above description, attached figures and their descriptions are intended to be illustrative and not limiting of this invention. and variations are within the contemplation hereof.

REFERENCES



[0072] 
  1. 1. Montecucco C & Schiavo G (1994) Mechanism of action of tetanus and botulinum neurotoxins. Mol Microbio113(1):1-8.
  2. 2. Poulain B & Humeau Y (2003) Mode of action of botulinum neurotoxin: pathological, cellular and molecular aspect. Ann Readapt Med Phys 46(6):265- 275.
  3. 3. Dong M, et al. (2003) Synaptotagmins I and II mediate entry of botulinum neurotoxin B into cells. J Cell Biol 162(7):1293-1303.
  4. 4. Dong M, et al. (2006) SV2 is the protein receptor for botulinum neurotoxin A. Science 312(5773):592-596.
  5. 5. Rummel A, Karnath T, Henke T, Bigalke H, & Binz T (2004) Synaptotagmins I and II act as nerve cell receptors for botulinum neurotoxin G. J Biol Chem 279(29):30865-30870.
  6. 6. Brunger AT (2005) Structure and function of SNARE and SNARE-interacting proteins. Q Rev Biophys 38(1):1-47.
  7. 7. Sadoul K, et al. (1997) SNAP-23 is not cleaved by botulinum neurotoxin E and can replace SNAP-25 in the process of insulin secretion. J Biol Chem 272(52):33023-33027.
  8. 8. Vaidyanathan VV, et al. (1999) Proteolysis of SNAP-25 isoforms by botulinum neurotoxin types A, C, and E: domains and amino acid residues controlling the formation of enzyme-substrate complexes and cleavage. J Neurochem 72(1):327-337.
  9. 9. Jahn R & Scheller RH (2006) SNAREs--engines for membrane fusion. Nat Rev Mol Cell Biol 7(9):631-643.
  10. 10. Mahajan ST & Brubaker L (2006) Botulinum toxin: From life-threatening disease to novel medical therapy. Am J Obstet Gynecol.
  11. 11. Mahajan ST & Brubaker L (2007) Botulinum toxin: from life-threatening disease to novel medical therapy. Am J Obstet GynecoI196(1):7-15.
  12. 12. Glogau RG (2002) Review of the use of botulinum toxin for hyperhidrosis and cosmetic purposes. Clin J Pain 18(6 Suppl):SI91-197.
  13. 13. Cheng CM, Chen JS, & Patel RP (2006) Unlabeled uses of botulinum toxins: a review, part 2. Am J Health Syst Pharm 63(3):225-232.
  14. 14. Chaddock JA, et al. (2004) Retargeted clostridial endopeptidases: inhibition of nociceptive neurotransmitter release in vitro, and antinociceptive activity in in vivo models of pain. Mov Disord 19 Supp18:S42-47.
  15. 15. Duggan MJ, et al. (2002) Inhibition of release of neurotransmitters from rat dorsal root ganglia by a novel conjugate of a Clostridium botulinum toxin A endopeptidase fragment and Erythrina cristagalli lectin. J Biol Chem 277(38):34846-34852.
  16. 16. Foster KA, et ai. (2006) Re-engineering the target specificity of Clostridial neurotoxins - a route to novel therapeutics. Neurotox Res 9(2-3): 101-107.
  17. 17. Davis CW & Dickey BF (2008) Regulated airway goblet cell mucin secretion. Annu Rev Physioi70:487-512.
  18. 18. Rogers DF (2007) Physiology of airway mucus secretion and pathophysiology of hypersecretion. Respir Care 52(9):1134-1146; discussion 1146-1139.
  19. 19. Chen S & Barbieri JT (2007) Multiple pocket recognition ofSNAP25 by botulinum neurotoxin serotype E. J Biol Chem 282(35):25540-25547.
  20. 20. Chen S & Barbieri JT (2006) Unique substrate recognition by botulinum neurotoxins serotypes A and E. J Biol Chem 281 (16): 10906-10911.
  21. 21. Sikorra S, Henke T, Galli T, & Binz T (2008) Substrate recognition mechanism of V AMP/synaptobrevin-cleaving clostridial neurotoxins. J Biol Chem 283(30):21145-21152.
  22. 22. Chen S, Hall C, & Barbieri JT (2008) Substrate recognition ofVAMP-2 by botulinum neurotoxin B and tetanus neurotoxin. J Biol Chem 283(30):21153-21159.
  23. 23. Oyler GA, et al. (1989) The identification of a novel synaptosomal-associated protein, SNAP-25, differentially expressed by neuronal subpopulations. J Cell Biol 109(6 Pt 1):3039-3052.
  24. 24. Holt M, et al. (2006) Identification of SNAP-47, a novel Qbc-SNARE with ubiquitous expression. J Biol Chem 281(25):17076-17083.
  25. 25. Pan PY, et al. (2005) SNAP-29-mediated modulation of synaptic transmission in cultured hippocampal neurons. J Biol Chem 280(27):25769-25779.
  26. 26. Okayama M, Arakawa T, Mizoguchi I, Tajima Y, & Takuma T (2007) SNAP-23 is not essential for constitutive exocytosis in HeLa cells. FEBS Lett 581(24):4583-4588.
  27. 27. Abonyo BO, et al. (2004) Syntaxin 2 and SNAP-23 are required for regulated surfactant secretion. Biochemistry 43(12):3499-3506.
  28. 28. Martin-Martin B, Nabokina SM, Blasi J, Lazo PA, & Mollinedo F (2000) Involvement of SNAP-23 and syntaxin 6 in human neutrophil exocytosis. Blood 96(7):2574-2583.
  29. 29. Pagan JK, et al. (2003) The t-SNARE syntaxin 4 is regulated during macrophage activation to function in membrane traffic and cytokine secretion. Curr Biol 13(2): 156-160.
  30. 30. Strom TB, et al. (1991) Immunotoxins and cytokine toxin fusion proteins. Ann N Y Acad Sci 636:233-250.
  31. 31. Du X, Ho M, & Pastan I (2007) New immunotoxins targeting CD123, a stem cell antigen on acute myeloid leukemia cells. J Immunother 30(6):607-613.
  32. 32. Baldwin MR & Barbieri JT (2007) Association of botulinum neurotoxin serotypes A and B with synaptic vesicle protein complexes. Biochemistry 46(11):3200-3210.
  33. 33. Washbourne P, et al. (2001) Cysteine residues of SNAP-25 are required for SNARE disassembly and exocytosis, but not for membrane targeting. Biochem J 357(Pt 3):625-634.
  34. 34. Winter, G. Harris, W. J. Humanized antibodies 1993, 14:6:243-6.
  35. 35. Nishibori, N.Horiuchi, H. Furusawa, S. Matsuda, H. Humanization of chicken monoclonal antibody using phage-display system. 2006, Mol Immunol, 43:6: 634-42.
  36. 36. Strom TB, et al. (1991) Immunotoxins and cytokine toxin fusion proteins. Ann N Y Acad Sci 636:233-250.
  37. 37. Du X, Ho M, & Pastan I (2007) New immunotoxins targeting CD123, a stem cell antigen on acute myeloid leukemia cells. J Immunother 30(6):607-613.
  38. 38. Milne JC, Blanke SR, Hanna PC, Collier RJ. (1995) Protective antigen-binding domain of anthrax lethal factor mediates translocation of a heterologous protein fused to its amino- or carboxy-terminus. Molecular Microbiology 15(4):661-6.
  39. 39. Haynes SM, Longmuir KJ, Robertson RT, Baratta JL, Waring AJ. Drug Deliv. (2008) 15(4):207-17.
  40. 40. Neurology. 2008 May 6;70(19):1699-706. Assessment: Botulinum neurotoxin for the treatment of movement disorders (an evidence-based review): report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology.
  41. 41. Proc Natl Acad Sci USA. 2009 106(23):9180-4. Engineering botulinum neurotoxin to extend therapeutic intervention. Barbeiri et al.
  42. 42. J Clin Oncol. 2009, 27(18):2983-90. Phase II trial of recombinant immunotoxin RFB4(dsFv)-PE38 (BL22) in patients with hairy cell leukemia.
  43. 43. Int J Pharm. 2000 Apr 25;200(1):27-39. Novel therapeutic nano-particles (lipocores): trapping poorly water soluble compounds.

SEQUENCE LISTING



[0073] 

<110> Medical college of Wisconsin

<120> ENGINEERED BOTULINUM NEUROTOXIN

<130> 650053.00177

<160> 8

<170> PatentIn version 3.5

<210> 1
<211> 4017
<212> DNA
<213> Clostridium botulinum

<400> 1





<210> 2
<211> 636
<212> DNA
<213> Homo sapiens

<400> 2

<210> 3
<211> 2053
<212> DNA
<213> Homo sapiens

<400> 3





<210> 4
<211> 1019
<212> DNA
<213> Homo sapiens

<400> 4

<210> 5
<211> 1925
<212> DNA
<213> Homo sapiens

<400> 5



<210> 6
<211> 28
<212> PRT
<213> Homo sapiens

<400> 6

<210> 7
<211> 1250
<212> PRT
<213> Clostridium botulinum

<400> 7











<210> 8
<211> 31
<212> PRT
<213> Clostridium botulinum

<400> 8




Claims

1. A modified BoNT/E catalytic domain, wherein the lysine residue located at:

(i) BoNT/E light chain position 224 of GenBank accession number X62089,or

(ii) a BoNT/E light chain residue corresponding to lysine 224 of GenBank accession number X62089
has been altered to be another amino acid;

and wherein said modified BoNT/E catalytic domain cleaves human SNAP23.
 
2. A modified BoNT/E catalytic domain according to Claim 1, wherein an exemplary sequence of said BoNT/E light chain is the sequence of botulinum toxin E subtype Beluga light chain deposited at GenBank with accession number X62089.
 
3. The modified catalytic domain of Claim 1, wherein said light chain position 224 or residue corresponding to lysine 224 has been altered to be aspartic acid, glutamic acid, or alanine.
 
4. The modified catalytic domain of Claim 1, wherein said light chain residue has been altered to be aspartic acid, and wherein the catalytic domain cleaves SNAP25 but does not cleave SNAP29 or SNAP47.
 
5. The modified catalytic domain of Claim 1, wherein an exemplary sequence of said human SNAP23 is the sequence of SNAP23 deposited at GenBank with accession number CR457212.
 
6. The modified catalytic domain of any preceding claim, additionally comprising a target molecule useful in a protein delivery system.
 
7. An engineered botulinum neurotoxin E or botulinum neurotoxin light chain or botulinum toxin catalytic domain which comprises the modified catalytic domain of any one of the preceding claims.
 
8. The modified catalytic domain of any one of Claims 1 to 6 or the engineered botulinum neurotoxin E, botulinum neurotoxin light chain or botulinum toxin catalytic domain of Claim 7 for use in botulinum toxin therapy.
 
9. The modified catalytic domain, engineered botulinum neurotoxin E, botulinum neurotoxin light chain or botulinum toxin catalytic domain of Claim 8 for use according to Claim 8 in the treatment of asthma, CF, chronic obstructive pulmonary disease, gastric acid efflux and inflammation, immune disorders with a cytokine component or cancers with a cytokine component.
 
10. A DNA sequence encoding the modified catalytic domain of any one of Claims 1 to 6 or the engineered botulinum neurotoxin E, botulinum neurotoxin light chain or botulinum toxin catalytic domain of Claim 7.
 


Ansprüche

1. Eine modifizierte BoNT/E-katalytische Domäne, wobei der Lysinrest, der sich an

(i) der BoNT/E-Leichtkettenposition 224 der GenBank-Zugangsnummer X62089 befindet, oder

(ii) ein BoNT/E-Leichtkettenrest, der Lysin 224 der GenBank-Zugangsnummer X62089 entspricht
geändert worden ist, so dass er eine andere Aminosäure ist;
und wobei die modifizierte BoNT/E-katalytische Domäne humanes SNAP23 spaltet.


 
2. Modifizierte BoNT/E-katalytische Domäne gemäß Anspruch 1, wobei eine beispielhafte Sequenz der BoNT/E-Leichtkette die bei GenBank unter der Zugangsnummer X62089 hinterlegte Sequenz der Botulinumtoxin-E-Subtyp-Beluga-Leichtkette ist.
 
3. Modifizierte katalytische Domäne gemäß Anspruch 1, wobei die Leichtkettenposition 224 oder der Rest, der Lysin 224 entspricht, so geändert worden ist, dass sie oder er Asparaginsäure, Glutaminsäure oder Alanin ist.
 
4. Modifizierte katalytische Domäne gemäß Anspruch 1, wobei der Leichtkettenrest so geändert worden ist, dass er Asparaginsäure ist, und wobei die katalytische Domäne SNAP25 spaltet, aber nicht SNAP29 oder SNAP47 spaltet.
 
5. Modifizierte katalytische Domäne gemäß Anspruch 1, wobei eine beispielhafte Sequenz des humanen SNAP23 die bei GenBank unter der Zugangsnummer CR457212 hinterlegte Sequenz von SNAP23 ist.
 
6. Modifizierte katalytische Domäne gemäß einem vorhergehenden Anspruch, die zusätzlich ein Target-Molekül, das in einem Proteinabgabesystem von Nutzen ist, beinhaltet.
 
7. Ein gentechnisch hergestelltes Botulinumneurotoxin E oder eine Botuliniumneurotoxin-Leichtkette oder eine katalytische Botulinumtoxin-Domäne, die die modifizierte katalytische Domäne gemäß einem der vorhergehenden Ansprüche beinhaltet.
 
8. Modifizierte katalytische Domäne gemäß einem der Ansprüche 1 bis 6 oder das gentechnisch hergestellte Botulinumneurotoxin E, die Botulinumneurotoxin-Leichtkette oder die katalytische Botulinumtoxin-Domäne gemäß Anspruch 7 zur Verwendung bei der Botulinumtoxintherapie.
 
9. Modifizierte katalytische Domäne, gentechnisch hergestelltes Botulinumneurotoxin E, Botulinumneurotoxin-Leichtkette oder katalytische Botulinumtoxin-Domäne gemäß Anspruch 8 zur Verwendung gemäß Anspruch 8 bei der Behandlung von Asthma, CF, chronisch obstruktiver Lungenerkrankung, Magensäureausfluss und -entzündung, Immunstörungen mit einer Zytokinkomponente oder Krebs mit einer Zytokinkomponente.
 
10. DNA-Sequenz, die für die modifizierte katalytische Domäne gemäß einem der Ansprüche 1 bis 6 oder für das das gentechnisch hergestellte Botulinumneurotoxin E, die Botulinumneurotoxin-Leichtkette oder die katalytische Botulinumtoxin-Domäne gemäß Anspruch 7 codiert.
 


Revendications

1. Domaine catalytique de BoNT/E modifié, dans lequel le résidu lysine situé aux emplacements :

(i) position 224 sur la chaîne légère de BoNT/E ayant le numéro d'accession GenBank X62089, ou

(ii) un résidu de chaîne légère de BoNT/E correspondant à la lysine 224 ayant le numéro d'accession GenBank X62089
a été modifié pour devenir un autre acide aminé ;
et ledit domaine catalytique de BoNT/E modifié clivant la SNAP23 humaine.


 
2. Domaine catalytique de BoNT/E modifié selon la revendication 1, dans lequel une séquence exemplaire de ladite chaîne légère de BoNT/E est la séquence de chaîne légère de la toxine botulique E sous-type Beluga déposée chez GenBank sous le numéro d'accession X62089.
 
3. Domaine catalytique modifié selon la revendication 1, dans lequel ladite position 224 sur la chaîne légère ou le résidu correspondant à la lysine 224 a été modifié(e) pour devenir l'acide aspartique, l'acide glutamique, ou l'alanine.
 
4. Domaine catalytique modifié selon la revendication 1, dans lequel ledit résidu de chaîne légère a été modifié pour devenir l'acide aspartique, et le domaine catalytique clivant SNAP25, mais ne clivant pas SNAP29 ou SNAP47.
 
5. Domaine catalytique modifié selon la revendication 1, dans lequel une séquence exemplaire de ladite SNAP23 humaine est la séquence de SNAP23 déposée chez GenBank sous le numéro d'accession CR457212.
 
6. Domaine catalytique modifié selon l'une quelconque des revendications précédentes, comprenant en outre une molécule cible qui peut être utilisée dans un système d'apport de protéines.
 
7. Neurotoxine botulique E ayant fait l'objet d'une manipulation ou chaîne légère de neurotoxine botulique ou domaine catalytique de toxine botulique qui comprend le domaine catalytique modifié selon l'une quelconque des revendications précédentes.
 
8. Domaine catalytique modifié selon l'une quelconque des revendications 1 à 6 ou neurotoxine botulique E ayant fait l'objet d'une manipulation, chaîne légère de neurotoxine botulique ou domaine catalytique de toxine botulique selon la revendication 7, destiné(e) à une utilisation dans une thérapie utilisant une toxine botulique.
 
9. Domaine catalytique modifié, neurotoxine botulique E ayant fait l'objet d'une manipulation, chaîne légère de neurotoxine botulique ou domaine catalytique de toxine botulique selon la revendication 8, destiné(e) à une utilisation selon la revendication 8 dans le traitement de l'asthme, de la mucoviscidose, de la bronchopneumopathie chronique obstructive, d'un reflux gastrique acide et d'une inflammation gastrique, d'affections immunes ayant une composante cytokine, ou de cancers ayant une composante cytokine.
 
10. Séquence d'ADN codant le domaine catalytique modifié selon l'une quelconque des revendications 1 à 6, ou neurotoxine botulique E ayant fait l'objet d'une manipulation, chaîne légère de neurotoxine botulique ou domaine catalytique de toxine botulique selon la revendication 7.
 




Drawing























Cited references

REFERENCES CITED IN THE DESCRIPTION



This list of references cited by the applicant is for the reader's convenience only. It does not form part of the European patent document. Even though great care has been taken in compiling the references, errors or omissions cannot be excluded and the EPO disclaims all liability in this regard.

Patent documents cited in the description




Non-patent literature cited in the description